Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Pediatr Res ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251881

ABSTRACT

Despite advances in neonatal care, the incidence of Bronchopulmonary Dysplasia (BPD) remains high among extreme preterm infants. The pathogenesis of BPD is multifactorial, with inflammation playing a central role. There is strong evidence that stem cell therapy reduces inflammatory changes and restores normal lung morphology in animal models of hyperoxia-induced lung injury. These therapeutic effects occur without significant engraftment of the stem cells in the host lung, suggesting more of a paracrine mechanism mediated by their secretome. In addition, there are multiple concerns with stem cell therapy which may be alleviated by administering only the effective vesicles instead of the cells themselves. Extracellular vesicles (EVs) are cell-derived components secreted by most eukaryotic cells. They can deliver their bioactive cargo (mRNAs, microRNAs, proteins, growth factors) to recipient cells, which makes them a potential therapeutic vehicle in many diseases, including BPD. The following review will highlight recent studies that investigate the effectiveness of EVs derived from stem cells in preventing or repairing injury in the preterm lung, and the potential mechanisms of action that have been proposed. Current limitations will also be discussed as well as suggestions for advancing the field and easing the transition towards clinical translation in evolving or established BPD. IMPACT: Extracellular vesicles (EVs) derived from stem cells are a potential intervention for neonatal lung diseases. Their use might alleviate the safety concerns associated with stem cell therapy. This review highlights recent studies that investigate the effectiveness of stem cell-derived EVs in preclinical models of bronchopulmonary dysplasia. It adds to the existing literature by elaborating on the challenges associated with EV research. It also provides suggestions to advance the field and ease the transition towards clinical applications. Optimizing EV research could ultimately improve the quality of life of extreme preterm infants born at vulnerable stages of lung development.

2.
Am J Physiol Lung Cell Mol Physiol ; 320(6): L1101-L1117, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33851545

ABSTRACT

Lung transplantation remains the only viable option for individuals suffering from end-stage lung failure. However, a number of current limitations exist including a continuing shortage of suitable donor lungs and immune rejection following transplantation. To address these concerns, engineering a decellularized biocompatible lung scaffold from cadavers reseeded with autologous lung cells to promote tissue regeneration is being explored. Proof-of-concept transplantation of these bioengineered lungs into animal models has been accomplished. However, these lungs were incompletely recellularized with resulting epithelial and endothelial leakage and insufficient basement membrane integrity. Failure to repopulate lung scaffolds with all of the distinct cell populations necessary for proper function remains a significant hurdle for the progression of current engineering approaches and precludes clinical translation. Advancements in 3D bioprinting, lung organoid models, and microfluidic device and bioreactor development have enhanced our knowledge of pulmonary lung development, as well as important cell-cell and cell-matrix interactions, all of which will help in the path to a bioengineered transplantable lung. However, a significant gap in knowledge of the spatiotemporal interactions between cell populations as well as relative quantities and localization within each compartment of the lung necessary for its proper growth and function remains. This review will provide an update on cells currently used for reseeding decellularized scaffolds with outcomes of recent lung engineering attempts. Focus will then be on how data obtained from advanced single-cell analyses, coupled with multiomics approaches and high-resolution 3D imaging, can guide current lung bioengineering efforts for the development of fully functional, transplantable lungs.


Subject(s)
Bioengineering , Lung Transplantation , Lung , Tissue Scaffolds , Animals , Bioengineering/methods , Extracellular Matrix/immunology , Humans , Lung/immunology , Lung/pathology , Lung Transplantation/methods , Tissue Engineering/methods
3.
Differentiation ; 105: 45-53, 2019.
Article in English | MEDLINE | ID: mdl-30711828

ABSTRACT

Considerable work has gone into creating cell therapies from induced pluripotent stem cells (iPSCs) since their discovery just over a decade ago. However, comparatively little research has been done concerning the safety of iPSCs and their progeny and specifically the mechanisms governing teratogenicity. The aim of this study was to ascertain at what developmental phase iPSCs undergoing differentiation to an alveolar-like phenotype lose their capacity to form a teratoma and uncover potential mechanisms responsible. iPSCs were differentiated using a previously published directed differentiation protocol mirroring alveolar embryogenesis. At each developmental phase cell phenotype was assessed and cells mixed with Matrigel and injected subcutaneously above the hind limbs of NSG mice to determine teratogenicity. A genetic screen of 42 genes commonly associated with teratoma formation was conducted on all the cells and any resulting teratoma. It was found that neither NKX2-1 lung progenitors nor terminally differentiated alveolar-like cells formed teratomas. As expected the expression of pluripotency markers was diminished over differentiation. However, the expression of two proteoglycans, decorin and lumican, was increased more than 3000x during differentiation. Both decorin and lumican are putative tumor suppressors with additional functions in angiogenesis, fibrosis, inflammation and autophagy. We hypothesize that the increasing expression of these proteoglycans by iPSCs as they differentiate may act to inhibit host endothelial cell recruitment when implanted resulting in the inhibition of any teratoma formation by any remaining undifferentiated iPSCs.


Subject(s)
Alveolar Epithelial Cells/cytology , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Phenotype , Stem Cell Transplantation/adverse effects , Teratoma/etiology , Animals , Cells, Cultured , Decorin/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Lumican/metabolism , Male , Mice , Proteoglycans/metabolism , Thyroid Nuclear Factor 1/metabolism
4.
Anal Chem ; 91(5): 3405-3411, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30741527

ABSTRACT

CD8+ T cells constitute an essential compartment of the adaptive immune system. During immune responses, naïve T cells become functional, as they are primed with their cognate determinants by the antigen presenting cells. Current methods of identifying activated CD8+ T cells are laborious, time-consuming and expensive due to the extensive list of required reagents. Here, we demonstrate an optical imaging approach featuring quantitative phase imaging to distinguish activated CD8+ T cells from naïve CD8+ T cells in a rapid and reagent-free manner. We measured the dry mass of live cells and employed transport-based morphometry to better understand their differential morphological attributes. Our results reveal that, upon activation, the dry cell mass of T cells increases significantly in comparison to that of unstimulated cells. By employing deep learning formalism, we are able to accurately predict the population ratios of unknown mixed population based on the acquired quantitative phase images. We envision that, with further refinement, this label-free method of T cell phenotyping will lead to a rapid and cost-effective platform for assaying T cell responses to candidate antigens in the near future.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Deep Learning , Humans , Microscopy, Phase-Contrast
5.
Pediatr Res ; 84(2): 306-313, 2018 08.
Article in English | MEDLINE | ID: mdl-29895838

ABSTRACT

BACKGROUND: The role of epithelial cells in eosinophilic esophagitis (EoE) is not well understood. In this study, our aim was to isolate, culture, and expand esophageal epithelial cells obtained from patients with or without EoE and characterize differences observed over time in culture. METHODS: Biopsies were obtained at the time of endoscopy from children with EoE or suspected to have EoE. We established patient-derived esophageal epithelial cell (PDEEC) lines utilizing conditional reprogramming methods. We determined integrin profiles, gene expression, MHC class II expression, and reactivity to antigen stimulation. RESULTS: The PDEECs were found to maintain their phenotype over several passages. There were differences in integrin profiles and gene expression levels in EoE-Active compared to normal controls and EoE-Remission patients. Once stimulated with antigens, PDEECs express MHC class II molecules on their surface, and when co-cultured with autologous T-cells, there is increased IL-6 and TNF-α secretion in EoE-Active patients vs. controls. CONCLUSION: We are able to isolate, culture, and expand esophageal epithelial cells from pediatric patients with and without EoE. Once stimulated with antigens, these cells express MHC class II molecules and behave as non-professional antigen-presenting cells. This method will help us in developing an ex vivo, individualized, patient-specific model for diagnostic testing for causative antigens.


Subject(s)
Eosinophilic Esophagitis/diagnosis , Epithelial Cells/metabolism , Esophagus/cytology , 3T3 Cells , Adolescent , Animals , Biopsy , Child , Child, Preschool , Endoscopy , Eosinophilic Esophagitis/metabolism , Gene Expression Profiling , Histocompatibility Antigens Class II/metabolism , Humans , Inflammation , Integrins/metabolism , Mice , T-Lymphocytes/metabolism
6.
Differentiation ; 96: 62-69, 2017.
Article in English | MEDLINE | ID: mdl-28802115

ABSTRACT

Protocols to differentiate induced pluripotent stem cells (iPSCs) into specialized cells are continually evolving. iPSCs can be differentiated to alveolar cells with protocols that focus on development, specifically by inducing differentiation into definitive endoderm (DE), anterior foregut endoderm (AFE) and then lung bud progenitor intermediaries. However, current protocols result in a relatively low yield of the desired alveolar cells. The aim of this study was to evaluate whether depleting uncommitted cells after AFE induction would have a beneficial effect on alveolar cell yield. iPSCs were differentiated on Matrigel-coated plates for 25days. At each stage, phenotype was assessed using flow cytometry, immunofluorescence and qRT-PCR. Additionally, samples were dissociated in trypsin following AFE induction to improve the purity of the cells for the subsequent lung differentiation phase. Finally, the efficacy of dissociating the samples was confirmed comparing the expression of markers indicative of pluripotency and apoptosis. The ability to differentiate iPSCs to DE was 96% and to AFE was 97% utilizing our current protocol. After depletion of uncommitted cells and 12 days in culture, the purity of lung bud progenitors was 99%. Finally, the percentage of alveolar types I and II at the end of differentiation was 74% as compared to 31% in control cultures that had not been depleted of uncommitted cells after AFE induction. In conclusion, depletion of uncommitted cells after AFE induction, improves terminal differentiation of alveolar cells from 31% to 74%.


Subject(s)
Alveolar Epithelial Cells/cytology , Cell Differentiation , Cellular Reprogramming Techniques/methods , Endoderm/cytology , Induced Pluripotent Stem Cells/cytology , Alveolar Epithelial Cells/metabolism , Cells, Cultured , Embryonic Induction , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism
7.
Adv Neonatal Care ; 16(6): 410-419, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27749687

ABSTRACT

BACKGROUND: The benefits of breast milk are well described, yet the mechanistic details related to how breast milk protects against acute and chronic diseases and optimizes neurodevelopment remain largely unknown. Recently, breast milk was found to contain stem cells that are thought to be involved in infant development. PURPOSE: The purpose of this review was to synthesize all available research involving the characterization of breast milk stem cells to provide a basis of understanding for what is known and what still needs further exploration. METHODS/SEARCH STRATEGY: The literature search was conducted between August and October 2015 using the CINAHL, PubMed, and reference list searching. Nine studies addressed characterization of human breast milk stem cells. FINDINGS/RESULTS: Five research teams in 4 countries have published studies on breast milk stem cells. Current research has focused on characterizing stem cells in full-term breast milk. The amount, phenotype, and expression of breast milk stem cells are known to vary between mothers, and they have been able to differentiate into all 3 germ layers (expressing pluripotent characteristics). IMPLICATIONS FOR PRACTICE: There is much to learn about breast milk stem cells. Given the potential impact of this research, healthcare professionals should be aware of their presence and ongoing research to determine benefits for infants. IMPLICATIONS FOR RESEARCH: Extensive research is needed to further characterize stem cells in breast milk (full-term and preterm), throughout the stages of lactation, and most importantly, their role in the health of infants, and potential for use in regenerative therapies.


Subject(s)
Gene Expression , Milk, Human/cytology , Pluripotent Stem Cells/cytology , Humans , Infant, Newborn , Infant, Premature , Milk, Human/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/physiology
9.
Stem Cell Res Ther ; 15(1): 80, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38486338

ABSTRACT

BACKGROUND: Despite advances in neonatal care, the incidence of Bronchopulmonary Dysplasia (BPD) remains high among preterm infants. Human induced pluripotent stem cells (hiPSCs) have shown promise in repairing injury in animal BPD models. Evidence suggests they exert their effects via paracrine mechanisms. We aim herein to assess the effectiveness of extracellular vesicles (EVs) derived from hiPSCs and their alveolar progenies (diPSCs) in attenuating hyperoxic injury in a preterm lung explant model. METHODS: Murine lung lobes were harvested on embryonic day 17.5 and maintained in air-liquid interface. Following exposure to 95% O2 for 24 h, media was supplemented with 5 × 106 particles/mL of EVs isolated from hiPSCs or diPSCs by size-exclusion chromatography. On day 3, explants were assessed using Hematoxylin-Eosin staining with mean linear intercept (MLI) measurements, immunohistochemistry, VEGFa and antioxidant gene expression. Statistical analysis was conducted using one-way ANOVA and Multiple Comparison Test. EV proteomic profiling was performed, and annotations focused on alveolarization and angiogenesis signaling pathways, as well as anti-inflammatory, anti-oxidant, and regenerative pathways. RESULTS: Exposure of fetal lung explants to hyperoxia induced airspace enlargement, increased MLI, upregulation of anti-oxidants Prdx5 and Nfe2l2 with decreased VEGFa expression. Treatment with hiPSC-EVs improved parenchymal histologic changes. No overt changes in vasculature structure were observed on immunohistochemistry in our in vitro model. However, VEGFa and anti-oxidant genes were upregulated with diPSC-EVs, suggesting a pro-angiogenic and cytoprotective potential. EV proteomic analysis provided new insights in regard to potential pathways influencing lung regeneration. CONCLUSION: This proof-of-concept in vitro study reveals a potential role for hiPSC- and diPSC-EVs in attenuating lung changes associated with prematurity and oxygen exposure. Our findings pave the way for a novel cell free approach to prevent and/or treat BPD, and ultimately reduce the global burden of the disease.


Subject(s)
Bronchopulmonary Dysplasia , Extracellular Vesicles , Hyperoxia , Induced Pluripotent Stem Cells , Lung Injury , Animals , Mice , Humans , Infant, Newborn , Hyperoxia/complications , Hyperoxia/metabolism , Hyperoxia/pathology , Animals, Newborn , Induced Pluripotent Stem Cells/metabolism , Lung Injury/therapy , Lung Injury/etiology , Antioxidants/metabolism , Proteomics , Infant, Premature , Lung/pathology , Bronchopulmonary Dysplasia/therapy , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Extracellular Vesicles/metabolism
10.
J Pediatr Surg ; 59(7): 1394-1398, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38614945

ABSTRACT

OBJECTIVE: The McGill Thyroid Nodule Score (MTNS) is a preoperative tool used to predict the risk for well-differentiated thyroid cancer in adults. It was developed by a multidisciplinary team using established evidence-based risk factors for thyroid cancer. The modified McGill Thyroid Nodule Score (mMTNS) was developed to predict malignancy risk in children. A pilot study suggested the mMTNS was able to assess malignancy risk in children with indeterminate cytology on fine needle aspiration (FNA). This study seeks to validate these findings. METHODS: Retrospective chart review identified subjects who underwent FNA biopsy and subsequent resection. Each patient was assigned a score to compare to final pathology. Statistical analysis was performed with SPSS. All tests were 2-tailed and statistical significance defined p < 0.05. Logistic regression used to determine predictive values of scores. RESULTS: 46 patients ≤21 years of age underwent resection of a thyroid nodule. Female predominance of 85% (n = 39). 78% (n = 36) of patients had palpable nodule. 65% (n = 30) found to have benign pathology and 35% (n = 16) found to have malignancy. Malignant nodules associated with greater mean mMTNS compared to benign [13.63 vs 7.23]. An mMTNS greater >12 had sensitivity of 86.7%, specificity of 90.3%, positive predictive value of 81.3%, and negative predictive value of 93.3%. CONCLUSION: Our data suggests the mMTNS continues to be a useful adjunct in predicting malignancy risk of pediatric thyroid nodules. An mMTNS >12 has a high risk for malignancy, which can aid in counseling and clinical decision making, particularly when there is indeterminate cytology on FNA. LEVEL OF EVIDENCE: IV.


Subject(s)
Thyroid Neoplasms , Thyroid Nodule , Humans , Thyroid Nodule/pathology , Thyroid Nodule/surgery , Female , Male , Child , Retrospective Studies , Thyroid Neoplasms/pathology , Thyroid Neoplasms/surgery , Adolescent , Risk Assessment/methods , Biopsy, Fine-Needle , Thyroidectomy , Young Adult , Risk Factors , Sensitivity and Specificity , Child, Preschool
11.
Respirology ; 18(6): 895-911, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23614471

ABSTRACT

For patients with end-stage lung diseases, lung transplantation is the only available therapeutic option. However, the number of suitable donor lungs is insufficient and lung transplants are complicated by significant graft failure and complications of immunosuppressive regimens. An alternative to classic organ replacement is desperately needed. Engineering of bioartificial organs using either natural or synthetic scaffolds is an exciting new potential option for generation of functional pulmonary tissue for human clinical application. Natural organ scaffolds can be generated by decellularization of native tissues; these acellular scaffolds retain the native organ ultrastructure and can be seeded with autologous cells towards the goal of regenerating functional tissues. Several decellularization strategies have been employed for lungs; however, there is no consensus on the optimal approach. A variety of cell types have been investigated as potential candidates for effective recellularization of acellular lung scaffolds. Candidate cells that might be best utilized are those which can be easily and reproducibly isolated, expanded in vitro, seeded onto decellularized matrices, induced to differentiate into pulmonary lineage cells, and which survive to functional maturity. Whole lung cell suspensions, endogenous progenitor cells, embryonic and adult stem cells and induced pluripotent stem (iPS) cells have been investigated for their applicability to repopulate acellular lung matrices. Ideally, patient-derived autologous cells would be used for lung recellularization as they have the potential to reduce the need for post-transplant immunosuppression. Several studies have performed transplantation of rudimentary bioengineered lung scaffolds in animal models with limited, short-term functionality but much further study is needed.


Subject(s)
Bioengineering/methods , Lung , Stem Cells , Tissue Scaffolds , Animals , Humans , Lung Transplantation , Models, Animal
12.
Methods Protoc ; 6(4)2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37489431

ABSTRACT

Urethral healing is plagued by strictures, impacting quality of life and medical costs. Various growth factors (GFs) have shown promise as therapeutic approaches to improve healing, but there is no protocol for in vitro comparison between GFs. This study focuses the development of a biomimetic in vitro urothelial healing assay designed to mimic early in vivo healing, followed by an evaluation of urothelial cell growth in response to GFs. METHODS: Wound-healing assays were developed with human urothelial cells and used to compared six GFs (EGF, FGF-2, IGF-1, PDGF, TGF-ß1, and VEGF) at three concentrations (1 ng/mL, 10 ng/mL, and 100 ng/mL) over a 48 h period. A commercial GF-containing medium (EGF, TGF-α, KGF, and Extract P) and a GF-free medium were used as controls. RESULTS: There was a statistically significant increase in cell growth for IGF-1 at 10 and 100 ng/mL compared to both controls (p < 0.05). There was a statistically significant increase in cell growth for EGF at all concentrations compared to the GF-free medium control (p < 0.05). CONCLUSION: This study shows the development of a clinically relevant wound-healing assay to evaluate urothelial cell growth. It is the first to compare GFs for future use in reconstructive techniques to improve urethral healing.

13.
J Surg Educ ; 80(1): 62-71, 2023 01.
Article in English | MEDLINE | ID: mdl-36085115

ABSTRACT

OBJECTIVE: The first transition to fellowship course for incoming pediatric surgery fellows was held in the US in 2018 and the second in 2019. The course aimed to facilitate a successful transition in to fellowship by introduction of the professional, patient care, and technical aspects unique to pediatric surgery training. The purpose of this study was to evaluate the feasibility and effectiveness of the first two years of this course in the US and discuss subsequent evolution of this endeavor. DESIGN: This is a descriptive and qualitative analysis of two years' experience with the Association of Pediatric Surgery Training Program Directors' (APSTPD) Transition to Fellowship course. Course development and curriculum, including clinical knowledge, soft skills, and hands-on skills labs, are presented. Participating incoming fellows completed multiple choice, boards-style pre- and post-tests. Scores were compared to determine if knowledge was effectively transferred. Participants also completed post-course evaluations and subsequent 3- or 12-month surveys inquiring on the lasting impact of the course on their transition into fellowship. Standard univariate statistics were used to present results. SETTING: The first APSTPD Transition to Fellowship course was held at the Johns Hopkins Hospital in Baltimore, Maryland in 2018, and the second course was held at the Oregon Health and Science University in Portland, Oregon in 2019. PARTICIPANTS: All fellows entering ACGME-certified Pediatric Surgery fellowships in the United States were invited to participate. Twenty fellows accepted and attended in 2018, and fourteen fellows participated in 2019. RESULTS: There were 34 incoming pediatric surgery fellow participants over 2 years. Faculty represented more than 10 institutions each year. Pre- and post-test scores were similar between years, with a significant improvement of scores after completion of the course (67±10% vs 79±8%, p < 0.001). Feedback from participants was overwhelmingly positive, with skills labs being attendees' favorite component. When asked about usefulness of individual course sessions, more attendees found clinical sessions more useful than soft skills (93% vs 73%, p = 0.011). Almost all (90%) of participants reported the course met its stated purpose and would recommend the course to future fellows. This was further reflected on 3 and 12 month follow up surveys wherein 85% stated they found the course helpful during the first few months of fellowship and 90% would still recommend it. CONCLUSIONS: A transition to fellowship course in the US for incoming pediatric surgery fellows is logistically feasible, effective in transfer of knowledge, and highly regarded among attendees. Feedback from each course has been used to improve the subsequent courses, ensuring that it remains a valuable addition to pediatric surgical training in the US.


Subject(s)
Fellowships and Scholarships , Specialties, Surgical , Child , Humans , United States , Education, Medical, Graduate/methods , Curriculum , Oregon , Surveys and Questionnaires
14.
J Pediatr Surg ; 58(6): 1213-1218, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36931942

ABSTRACT

INTRODUCTION: Endoscopic surveillance guidelines for patients with repaired esophageal atresia (EA) rely primarily on expert opinion. Prior to embarking on a prospective EA surveillance registry, we sought to understand EA surveillance practices within the Eastern Pediatric Surgery Network (EPSN). METHODS: An anonymous, 23-question Qualtrics survey was emailed to 181 physicians (surgeons and gastroenterologists) at 19 member institutions. Likert scale questions gauged agreement with international EA surveillance guideline-derived statements. Multiple-choice questions assessed individual and institutional practices. RESULTS: The response rate was 77%. Most respondents (80%) strongly agree or agree that EA surveillance endoscopy should follow a set schedule, while only 36% claimed to perform routine upper GI endoscopy regardless of symptoms. Many institutions (77%) have an aerodigestive clinic, even if some lack a multi-disciplinary EA team. Most physicians (72%) expressed strong interest in helping develop evidence-based guidelines. CONCLUSIONS: Our survey reveals physician agreement with current guidelines but weak adherence. Surveillance methods vary greatly, underscoring the lack of evidence-based data to guide EA care. Aerodigestive clinics may help implement surveillance schedules. Respondents support evidence-based protocols, which bodes well for care standardization. Results will inform the first multi-institutional EA databases in the United States (US), which will be essential for evidence-based care. LEVEL OF EVIDENCE: This is a prognosis study with level 4 evidence.


Subject(s)
Esophageal Atresia , Tracheoesophageal Fistula , Child , Humans , Esophageal Atresia/surgery , Esophageal Atresia/epidemiology , Tracheoesophageal Fistula/surgery , Prospective Studies , Surveys and Questionnaires
15.
Conn Med ; 76(3): 171-2, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22666978

ABSTRACT

Pediatric surgical subspecialty workforce shortages are here to stay without any expected solution for the short-term. Individual surgeons, hospital administrators, risk management and patient-safety teams need to recognize that patient safety must take precedence over clinical productivity and financial "bottom lines." Pushing attending surgeon work hours beyond the limits of exhaustion impairs patient safety. Just as resident surgeon work hours have been appropriately curtailed in the name of patient safety, so must attending surgeon work hours. This issue needs to be addressed by hospital patient safety committees, professional societies, and by state and national regulating authorities.


Subject(s)
Medical Staff, Hospital , Patient Safety , Pediatrics , Sleep Deprivation , Work Schedule Tolerance , Child , Fatigue , General Surgery , Humans , Workload
16.
Conn Med ; 76(3): 159-62, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22666976

ABSTRACT

INTRODUCTION: Pediatric trauma requires several medical professionals working together as a highly functioning team. Since critical pediatric medical events are rare, routine practice can be one method of ensuring that all members communicate and provide quality care. The goal of this study was to evaluate whether high-fidelity simulation training improved surgery residents' knowledge and self-efficacy in relation to pediatric trauma scenarios. METHODS: Participants attended training that included initial assessments of knowledge and self-efficacy, lecture, and practice with three trauma scenarios. After the training, residents completed pretest measures again. RESULTS: Findings indicated that resident knowledge and self-efficacy significantly increased afterthe simulation training. CONCLUSION: The current study suggests that for the adequate care of pediatric patients during trauma/emergency situations, high-fidelity simulation training may be beneficial. This may be particularly true for emergencies that are less frequent and not well practiced by clinicians and students.


Subject(s)
General Surgery/education , Internship and Residency , Patient Simulation , Pediatrics/education , Teaching/methods , Child , Child, Preschool , Female , Health Knowledge, Attitudes, Practice , Humans , Infant , Learning , Male , Pilot Projects , Program Evaluation , Self Efficacy , Surveys and Questionnaires
17.
Genes (Basel) ; 13(10)2022 10 15.
Article in English | MEDLINE | ID: mdl-36292751

ABSTRACT

Obesity is a growing health problem that affects both children and adults. The increasing prevalence of childhood obesity is associated with comorbidities such as cardiovascular disease, type 2 diabetes and metabolic syndrome due to chronic low-grade inflammation present at early stages of the disease. In pediatric patients suffering from obesity, the role of epigenetics, the gut microbiome and intrauterine environment have emerged as causative factors Interestingly, pediatric obesity is strongly associated with low birth weight. Accelerated weight gain oftentimes occurs in these individuals during the post-natal period, which can lead to increased risk of adiposity and metabolic disease. The pathophysiology of obesity is complex and involves biological and physiological factors compounded by societal factors such as family and community. On a cellular level, adipocytes contained within adipose tissue become dysregulated and further contribute to development of comorbidities similar to those present in adults with obesity. This review provides an overview of the current understanding of adipose tissue immune, inflammatory and metabolic adaptation of the adipose tissue in obesity. Early cellular changes as well as the role of immune cells and inflammation on the progression of disease in pivotal pediatric clinical trials, adult studies and mouse models are emphasized. Understanding the initial molecular and cellular changes that occur during obesity can facilitate new and improved treatments aimed at early intervention and subsequent prevention of adulthood comorbidities.


Subject(s)
Diabetes Mellitus, Type 2 , Pediatric Obesity , Pediatrics , Mice , Animals , Child , Humans , Diabetes Mellitus, Type 2/metabolism , Pediatric Obesity/epidemiology , Pediatric Obesity/genetics , Adipose Tissue/metabolism , Inflammation/metabolism
18.
NPJ Regen Med ; 7(1): 1, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35013320

ABSTRACT

Diseases of the esophagus, damage of the esophagus due to injury or congenital defects during fetal esophageal development, i.e., esophageal atresia (EA), typically require surgical intervention to restore esophageal continuity. The development of tissue engineered tubular structures would improve the treatment options for these conditions by providing an alternative that is organ sparing and can be manufactured to fit the exact dimensions of the defect. An autologous tissue engineered Cellspan Esophageal ImplantTM (CEI) was surgically implanted into piglets that underwent surgical resection of the esophagus. Multiple survival time points, post-implantation, were analyzed histologically to understand the tissue architecture and time course of the regeneration process. In addition, we investigated CT imaging as an "in-life" monitoring protocol to assess tissue regeneration. We also utilized a clinically relevant animal management paradigm that was essential for long term survival. Following implantation, CT imaging revealed early tissue deposition and the formation of a contiguous tissue conduit. Endoscopic evaluation at multiple time points revealed complete epithelialization of the lumenal surface by day 90. Histologic evaluation at several necropsy time points, post-implantation, determined the time course of tissue regeneration and demonstrated that the tissue continues to remodel over the course of a 1-year survival time period, resulting in the development of esophageal structural features, including the mucosal epithelium, muscularis mucosae, lamina propria, as well as smooth muscle proliferation/migration initiating the formation of a laminated adventitia. Long term survival (1 year) demonstrated restoration of oral nutrition, normal animal growth and the overall safety of this treatment regimen.

19.
J Pediatr Surg ; 56(1): 17-25, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33121738

ABSTRACT

BACKGROUND: Preclinical studies demonstrate that tissue engineering and patient-derived stem cells can regenerate tissue. The goal of this study was to determine whether stem cells from esophageal atresia patients (EA) could be utilized for this purpose. METHODS: Adipose tissue was obtained from control, esophageal atresia (EA) and long gap esophageal atresia (LGEA) patients. Mesenchymal stem cells (MSCs) were isolated, expanded, characterized and seeded onto tubular scaffolds for 6 days. Scaffolds were characterized for viability, gene expression and cytokine production. RESULTS: The average weight of tissue from the EA and LGEA patients was 145.8mg compared to 2981 mg in controls. Despite the small amount of tissue obtained from neonatal patients, cells were expanded to cover a scaffold. After incubating 6 days on the scaffold, cells were viable and proliferating with differences in gene expression between groups. VEGFA production in the supernatant was increased in EA and LGEA patients; while IL6 production was significantly increased in the control patients. CONCLUSIONS: This study demonstrates the ability to utilize small amounts of adipose tissue from esophageal atresia patients as a cell source for regenerative medicine. Future studies will focus on use of these cells for tissue regeneration in vivo.


Subject(s)
Esophageal Atresia , Mesenchymal Stem Cells , Esophageal Atresia/surgery , Humans , Infant, Newborn , Regenerative Medicine , Tissue Engineering , Tissue Scaffolds , Wound Healing
20.
Dis Model Mech ; 14(2)2021 02 23.
Article in English | MEDLINE | ID: mdl-33729989

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a chronic lung disease caused by exposure to high levels of oxygen (hyperoxia) and is the most common complication that affects preterm newborns. At present, there is no cure for BPD. Infants can recover from BPD; however, they will suffer from significant morbidity into adulthood in the form of neurodevelopmental impairment, asthma and emphysematous changes of the lung. The development of hyperoxia-induced lung injury models in small and large animals to test potential treatments for BPD has shown some success, yet a lack of standardization in approaches and methods makes clinical translation difficult. In vitro models have also been developed to investigate the molecular pathways altered during BPD and to address the pitfalls associated with animal models. Preclinical studies have investigated the efficacy of stem cell-based therapies to improve lung morphology after damage. However, variability regarding the type of animal model and duration of hyperoxia to elicit damage exists in the literature. These models should be further developed and standardized, to cover the degree and duration of hyperoxia, type of animal model, and lung injury endpoint, to improve their translational relevance. The purpose of this Review is to highlight concerns associated with current animal models of hyperoxia-induced BPD and to show the potential of in vitro models to complement in vivo studies in the significant improvement to our understanding of BPD pathogenesis and treatment. The status of current stem cell therapies for treatment of BPD is also discussed. We offer suggestions to optimize models and therapeutic modalities for treatment of hyperoxia-induced lung damage in order to advance the standardization of procedures for clinical translation.


Subject(s)
Bronchopulmonary Dysplasia/metabolism , Hyperoxia , Amnion/metabolism , Animals , Animals, Newborn , Apoptosis , Disease Models, Animal , Disease Progression , Epithelial Cells/metabolism , Humans , Hyperoxia/metabolism , In Vitro Techniques , Infant, Newborn , Infant, Premature , Lung/pathology , Lung/physiopathology , Lung Injury/pathology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Oxygen/chemistry , Oxygen/metabolism , Rats , Stem Cells/cytology , Translational Research, Biomedical , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL